2023 BAARN Program

Stuary Levy Lecture

David C. Hooper

David C. Hooper, M.D.

Professor of Medicine, Harvard Medical School
Physician, Former Chief Infection Control, Former Associate Chief Division of Infectious Diseases, Massachusetts General Hospital

Dr. Hooper

he/him/his

Dr. Hooper received his BA in Microbiology from the University of Texas at Austin and his MD from Washington University School of Medicine, St. Louis, followed by residency in Internal Medicine and fellowship in Infectious Diseases at Massachusetts General Hospital. His research has focused on mechanisms of antimicrobial action and resistance and epidemiology of resistance and hospital infections. He is author of over 200 peer-reviewed articles on mechanisms and epidemiology of antimicrobial resistance and 76 book chapters and other articles. Dr. Hooper is a Fellow of the American Academy of Microbiology, the Infectious Diseases Society of America, and the American Association of the Advancement of Science and a Member of the Association of American Physicians. He has served as Chair of the Drug Discovery and Antimicrobial Resistance Study Section of the NIH and Chair of the Scientific Program Committee of the Interscience Conference on Antimicrobial Agents and Chemotherapy of the American Society for Microbiology (ASM). He is past President and past Meetings Board Chair of the ASM, has served on the editorial board of Antimicrobial Agents and Chemotherapy, and is past Deputy Editor of The Journal of Infectious Diseases.

Talk: Quinolone and Multidrug Resistance: The Challenge of Microbial Ingenuity

Stuart Levy was a pioneer in the field of antimicrobial resistance, being one of the first to identify the broader risk of indirect antibiotic use and resistance spread from animals to humans and to recognize the importance of antimicrobial stewardship, for which he became an unrelenting advocate. He was also among the first to identify resistance due to active efflux of tetracycline and other antimicrobials and then to evaluate the regulatory networks that modulated these resistance mechanisms and phenotypes.

  Fluoroquinolones are synthetic bactericidal antimicrobials that came into clinical use in the 1980s and were later found to have two essential topoisomerase targets, both features that were thought to contribute to a low risk of resistance development. They became widely used because of overall good tolerability and many approved clinical indications. Early members of the class were, however, found to have differential activity against their two targets enabling a stepwise pathway to increasing resistance, leading to dual target resistance mutations in clinical resistant isolates. Active efflux was also progressively recognized, as more chromosomally encoded bacterial pumps with broad transport profiles, some of which included synthetic fluoroquinolones, were found beyond the plasmid-encoded Tet pumps that Levy had uncovered. The broad substrate profiles of these pumps mechanistically linked fluoroquinolone resistance to multidrug resistance. It was also initially thought that plasmid-mediated transferable fluoroquinolone resistance was unlikely to occur due to the dominance of sensitivity over resistance in the presence of hetero-diploid target gene alleles. George Jacoby, while studying β-lactamase genes transferred on plasmids, however, identified co-transfer of fluoroquinolone resistance mediated by qnr genes, which encode pentapeptide repeat proteins that interact with and protect both topoisomerase targets from fluoroquinolone action. We later identified active site mutants of a common plasmid-encoded aminoglycoside acetyltransferase that acquired the ability to reduce ciprofloxacin activity by acetylation of its secondary piperazinyl amine. Both of these plasmid-mediated fluoroquinolone resistance determinants are now widespread in enteric pathogens worldwide and often on multidrug resistance plasmids, further linking fluoroquinolone resistance with multidrug resistance. The scope of microbial ingenuity in developing resistance to antimicrobials, even classes with promising “low-resistance-risk” properties, is a strong argument for the continuing need for attention to antimicrobial stewardship, infection prevention and control, monitoring and investigation of resistance mechanisms, and new drug development.

Advances in Antibiotics

Session Description

This session is co-chaired by Erin Duffy, PhD, Chief of Reserach and Development at CARB-X, and Kevin Outterson, Executive Director of CARB-X.

In lieu of individual talks, Dr. Duffy will moderate a panel discussion moderated on the topic Challenges and opportunities in developing a high-impact innovative therapeutic for AMR featuring three presenters from the CARB-X portfolio.

Jonathan Stekbeck

Jonathan Steckbeck, PhD

Founder & CEO, Peptilogics

Dr. Stekbeck

he/him/his

Jonathan Steckbeck, PhD, MBA, is the founder and CEO of Peptilogics, a clinical-stage biotechnology platform company pioneering the development of best-in-class novel therapeutics. Under his leadership, Peptilogics has developed a computational drug discovery platform to speed the design and development of novel drugs across a range of therapeutic targets. The company is advancing a pipeline of novel therapeutics in focused indications and continually expanding the application of its platform to new disease areas.

Product in development: PLG0206, an investigational antibiotic peptide candidate for the treatment of periprosthetic joint infection (PJI)

Veronica Hall

Veronica Hall, PhD

Chief Operating Officer, Clarametyx Biosciences

Dr. Hall

she/her/hers

Veronica Hall, PhD, has a multidisciplinary background with more than 20 years of experience in life science research, including conception, design, and delivery of technical and commercialization solutions for vaccines, therapeutics, and drug/device combinations. Over the course of her career, Dr. Hall has successfully led R&D and partnership efforts, leading teams large and small for a range of small to mid-size biotech firms. She served as Senior Director at Emergent BioSolutions Inc. leading the strategic R&D pipeline growth. Dr. Hall received a Bachelor of Science in Biochemistry and Molecular Biology from University of California, Santa Cruz, and a Ph.D. in Biochemistry and Cellular Biology from Rice University. She completed her postdoctoral fellowship at the National Cancer Institute.

Product in development: CMTX-101, an investigational non-antibiotic antibody targeting the scaffolding proteins in bacterial biofilms

Matthew Henn

Matthew Henn

EVP R&D, Chief Scientific Officer, Seres Therapeutics

Dr. Henn

he/him/his

Matthew Henn is Executive Vice President and Chief Scientific Officer at Seres Therapeutics. He has been involved in the discovery and development of multiple microbiome therapeutics across infectious, inflammatory, and oncology indications including VOWSTtm the first FDA-approved oral microbiota-based therapeutic. He has authored over 70 peer-reviewed publications; his research has focused on microbial populations and the functional role of microbes in both environmental and human disease applications, and also the development of genomic and functional tools to study these populations. Prior to helping launch Seres in 2012, he was the Director of Viral Genomics and Assistant Director of the Genome Sequencing Center for Infectious Diseases at the Broad Institute of MIT and Harvard. He has served on various NIH working groups on antimicrobial resistance and microbiome research, as a scientific advisor for the National Institutes of Health’s Viral Pathogen Bioinformatics Resource Center, as an ad-hoc reviewer and editor of various peer-reviewed journals, and as a scientific advisor to non-profit and for-profit organizations. He currently serves on the Board of Directors of the Microbiome Therapeutics Innovation Group and the scientific advisory board of Growcentia, Inc, an agricultural microbiome company. Dr. Henn is formally trained in ecology and evolutionary biology and earned his Ph.D. in ecosystem sciences from the University of California at Berkeley, where he was a NASA Earth Systems Sciences Fellow, and trained as a NSF Postdoctoral Fellow in Microbiology at Duke University.

Product in development: SER-155, an investigational, oral, rationally designed, fermented microbiome therapeutic designed to reduce the risk of gastrointestinal infections, bacteremia, and graft versus host disease (GvHD) in immunocompromised patients

Advances in Diagnostics

Jong Lee

Jong Lee, MBA

CEO & Cofounder, Day Zero Diagnostics

he/him/his

Mr. Jong Lee is a co-founder and the CEO of Day Zero Diagnostics (DZD). DZD is developing a new class of infectious disease diagnostics for an era of growing antimicrobial resistance. The diagnostic platform incorporates proprietary technologies for ultra-high enrichment sample prep and sequencing data analysis to provide same day organism ID and AMR profiling directly from native samples. The platform will enable physicians to avoid prolonged empiric therapy and to switch to a targeted antimicrobial treatment within 8 hours rather than 2-5 days.

Prior to DZD, Jong spent two decades in strategy consulting focused on medical technology innovation in diabetes health care, cardiology, gastroenterology, and blood banking. He most recently served as the Senior VP of Marketing and Strategy for an early stage orthopedic company in joint arthroplasty.

Talk: Rapid Sequencing-Based Organism Detection and Antimicrobial Susceptibility Profiling for Clinical Diagnostics

Sequencing-based diagnostics are an emerging method for infectious disease testing that leverage an agnostic, metagenomic approach for the identification of pathogens. However, there are barriers to the realization of sequencing-based diagnostics for clinical decision making, including separation of signal from causative pathogens versus spurious signals caused by background commensals as well as from the up to billion-fold excess of human DNA. 

Blood2Bac™ is a rapid, culture-free, pathogen-agnostic sample preparation strategy that enriches bacterial and fungal DNA by up to 10 logs relative to human DNA from patient whole blood samples. This ultra high enrichment enables rapid whole genome sequencing (WGS) and computational analysis with a machine learning-based software package called Keynome® to provide accurate species identification (ID) and antimicrobial susceptibility (AST) directly from whole blood at single digit CFU/mL pathogen concentrations in less than 8 hours.

The DZD assay demonstrates the ability to accurately identify a broad range of bacterial and fungal BSI pathogens at single digit CFU/mL concentrations, with data from 35 species included in the presentation. The majority of samples resulted in near complete genome recovery, with an average 1x coverage of 94.3% for bacteria and 99.3% for fungi.  We demonstrate a high specificity of 97.1% with just 3 false positive calls across 105 tested samples. AST testing was performed on 10 priority BSI strains, with a total of 109 AST predictions made on 47 individual bug/drug combinations.  Keynome gAST predictions were compared to phenotypic results and achieved 92.7% categorical agreement. Total turnaround time for Blood2Bac™ ultra high enrichment was an average of 5.77 hours and we previously demonstrated a singleplex time of a total of 2 hours for library preparation, ONT sequencing, and computational analysis.

The ability of Blood2Bac™ to rapidly recover whole pathogen genomes and for Keynome® to provide accurate ID and AST predictions demonstrates the potential for integration of rapid, sequencing based diagnostics into clinical management of BSIs in the near future.

Justin Rolando

Dr. Justin Rolando, PhD

Research Fellow, Brigham and Women’s Hospital - Department of Pathology; Postdoctoral Fellow, Wyss Institute for Biologically Inspired Engineering; Research Fellow, Harvard Medical School.

he/him/his

Dr. Rolando's research addresses a common challenge in human health: the inability to diagnose diseases early enough for effective treatment, especially when symptoms are latent or non-specific. Using liquid biopsy and longitudinal studies of human health, he seeks to diagnose nascent diseases. Specifically, using quantitative state-of-the-art single-molecule protein-based and nucleic acid-based diagnostic tools (e.g., SIMOA, LAMP, and CRISPR), predictive machine learning models, clinical imaging, and novel biomarkers, Justin uncovers the molecular mechanisms of how chemical heterogeneity leads to overarching organism-level phenomena like pediatric sepsis, cancer, and antimicrobial resistance. When developed further, the quantitative pathophysiological research from his teams identify fingerprints of disease, paving the way for reduced human morbidity and mortality through in vitro diagnostic systems.

Talk: Ultra-Sensitive Salivary Profiling: Insights into Infants Treated for Suspected Sepsis

Correctly identifying an infected infant to provide timely therapy without exposing a non-infected infant to unnecessary antibiotics remains a significant diagnostic challenge in neonatology. Early clinical symptoms of infection are subtle, nonspecific, or mirror symptoms commonly seen in the uninfected premature newborn. For this reason, “rule out sepsis” is one of the most common diagnostic tests in neonatal intensive care units. The Salivary Profiling for Infants Treated for Suspected Sepsis (SPITSS Trial) is an NICHD funded, ongoing, multi-center, prospective observational study aimed at determining the accuracy of an ultra-sensitive multiplexed salivary cytokine panel to correctly determine the infectious status of neonates using host-response biomarkers. This presentation will detail an interim analysis and the importance of ultra-sensitive measurements to show that neonatal saliva is a rich source of cytokines that can be noninvasively quantified to aid in clinical decision making.

Yonatan Grad

Yonatan Grad, MD, PhD

Professor of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health

Dr. Yonatan Grad

he/him/his

Yonatan Grad is Professor of Immunology and Infectious Diseases at the Harvard TH Chan School of Public Health, and a member of the Division of Infectious Diseases at Brigham and Women’s Hospital. The Grad laboratory uses interdisciplinary methods—including molecular microbiology, pathogen genomics, and mathematical modeling—to study how pathogens evolve and spread through populations, with the motivation of improving clinical and public health strategies to decrease the burden of disease. A major theme of the lab’s work is antimicrobial resistance, with a particular focus on Neisseria gonorrhoeae, the causative agent of the sexually transmitted disease gonorrhea.

Talk: Addressing AMR in Neisseria gonorrhoeae: the roles and challenges of diagnostics

Neisseria gonorrhoeae, the causative agent of the sexually transmitted disease gonorrhea, poses a global health threat with an increasing rate of infections and increasing antibiotic resistance. Timely and accurate diagnostics are paramount for effective management and control. However, most diagnosis is via nucleic acid amplification tests, which do not inform on antibiotic susceptibilities, and treatment is most commonly empiric ceftriaxone, the last line antibiotic. With strains now circulating globally that have reduced susceptibility to ceftriaxone and resistance or reduced susceptibility to all approved antibiotics, there is an urgent need for rapid diagnostics that can help tailor therapy. The microbial ecology and selective pressures acting on N. gonorrhoeae are also about to change with (1) the uptake of doxycycline post-exposure prophylaxis and (2) the recent report that a new antibiotic—zoliflodacin—successfully completed its Phase 3 trials for treatment of uncomplicated gonorrhea. In this talk, we will discuss the use of population genomics to help inform the development of new diagnostics, the potential impact of escape from sequence-based diagnostics for antibiotic susceptibility, and strategies around the implementation and monitoring for doxycycline post-exposure prophylaxis and zoliflodacin.

Alternative Approaches

Mihaela Gadjeva

Mihaela Gadjeva, PhD

Associate Director, Moderna Theraputics

she/her/hers

Dr. Gadjeva is an internationally recognized immunologist with research interests in host-bacterial pathogenesis. She has an undergraduate and graduate degrees in Biochemistry from Sofia University, Bulgaria, and Oxford University, UK. After her postdoctoral training at Brigham and Women’s Hospital (BWH), Harvard Medical School, she rose through the Faculty ranks to an Associate Professor and maintained an NIH-funded lab for the past 12 years. Recently, Dr. Gadjeva joined Moderna Therapeutics where she leads bacterial vaccinology efforts. Her group applies system bacteriology and immunology approaches utilizing multiplexed quantitative proteomic and spatial transcriptomic analysis with the primary goal to discover key factors that determine outcomes of disease. The objective is to leverage this knowledge to discover novel therapeutic approaches to strengthen immunity during bacterial infections. Dr. Gadjeva has published more than 60 peer-reviewed scientific manuscripts and review articles in journals such as Nature Communications, Plos Pathogens, Immunity, Frontiers in Immunology, NEJM, etc.  Dr. Gadjeva’s contributions have been recognized by multiple awards such as First Place ARVO-AFER Merck Innovation award, Complement Society award, FOCIS award, BWH Innovation Award, ets. She also serves as an adviser to the Biotechnology Graduate Degree program at Harvard University.

Talk: Towards generation of an integrated map of infection

As there is a strong need to develop intervention strategies against ESKAPE pathogens, there is a great interest in developing new approaches that will bring depth to our understanding of the biology of these pathogens. I will discuss our efforts at developing comparative genomics and transcriptomics approaches to generate unified host-pathogen maps of infection which will ultimately lead to intervention opportunities. Genomic data from P. aeruginosa and E. coli clinical isolates were collected from the past 20 years and analyzed using comparative genomics to elucidate changes in the core genomes and frequencies of AMR acquisition. The analysis pointed to a staggering 40% increase in the appearance of AMR resistance traits in the clinical strains, tendencies that necessitate rapid interventions. To gain insights into the pathogen behavior within the infected tissues, we developed spatial, pathogen-specific transcriptome profiling and found differential enrichment of pathogen-specific transcripts at distinct anatomical locations. By using machine learning approaches, we correlated pathogen transcriptional features to host responses and found interconnected circuits. Cumulatively, our data highlight coordinated spatial and temporal transcriptional interplay between host and that may pinpoint to druggable circuits. 

Jared Kehe

Jared Kehe, PhD

Co-founder & Chief Scientific Officer, Concerto Biosciences

A passionate biotechnologist with expertise in microbial ecology, microbiome science, and microfluidics, Jared is Co-founder and Chief Scientific Officer of Concerto Biosciences, a Boston-based startup that designs microbial products. Jared co-invented Concerto’s platform, kChip, a discovery engine that physically constructs millions of miniature, defined microbial communities simultaneously. By observing the behavior of these communities, Jared’s team rapidly identifies promising microbial, prebiotic, and postbiotic product candidates. Aiming to unleash a new era of microbial research, Jared oversees discovery projects spanning skin health, vaginal health, gut health, agriculture, and food science. Jared holds a PhD in Biological Engineering from MIT (NSF Fellow, Presidential Fellow), where he worked with Prof. Paul Blainey to develop kChip and apply it to foundational questions in microbial ecology.

Talk: Post-Pathogen: How Microbial Ecology-Based Interventions Can Pacify—Rather than Kill—Our Symbionts

Antibiotics, crucial for combatting life-threatening infections, ironically create the very pathogens we seek to eradicate: Beyond selecting for antimicrobial resistance, antibiotics kill virulence-suppressing commensal microorganisms and disrupt host immunity. The healthy microbiome, composed of the diverse microbes that co-evolved symbiotically alongside humans, placate would-be pathogens and resist their colonization. Designing antibiotic alternatives or infection preventatives that recreate this microbiome-endowed protection requires an unprecedented understanding of how microbes behave together. Concerto unlocks this knowledge with kChip, a discovery engine that physically constructs millions of miniature, defined microbial cocultures simultaneously. By observing the behavior of a pathogen within each coculture, Concerto rapidly identifies the most promising microbial combinations, prebiotics, and postbiotics for development into new products. In our first endeavor, Concerto constructed >6 million communities of skin-dwelling microbes to discover an “ensemble” of bacteria that pacifies pathogenic S. aureus. We have since initiated discovery projects in vaginal health, gut health, and agriculture. In partnership with a diverse array of biotech and biopharma companies, Concerto aims to vastly improve our understanding of microbial ecology and develop durable alternatives to traditional antibiotics.

Janira Prichula

Janira Prichula, PhD

Postdoctoral Research Fellow at Gilmore’s Lab, Harvard Medical School/ Massachusetts Eye and Ear

she/her/hers

I am a postdoctoral research fellow currently working in the Gilmore Lab, Harvard Medical School/ Massachusetts Eye and Ear, Boston, with interest in genomics related to antibiotic resistance. I had the opportunity to conduct research on the global distribution of enterococci and antibiotic resistance genes in marine environments in remote ecologies of Patagonia and other regions of South America during my Master's and Ph.D. research. These experiences have shown me that understanding how bacteria relate to their hosts, what traits are central to colonization, and determining where reservoirs of resistant strains occur will provide critical information for preventing and managing the problem of multidrug-resistant bacteria infection. Currently, I am studying new approaches for the control of enterococcal infection. 

Talk: Efagins – A New Class of Phage-derived Anti-enterococcal Agents

Enterococci are leading causes of multidrug-resistant hospital infections. Here we identify a new potentially therapeutically useful bactericidal agent for treating enterococcal infections we term efagins.  Efagins appear to represent a new type of anti-enterococcal entity that evolved from elements of phages, with antibacterial properties – E. faecalis phage-derived bacteriocins. They appear to be unique to the species E. faecalis, and are encoded as part of the core chromosome of that species. Their antibacterial property, however, extends to other enterococcal species and possibly broader. Clues to the mechanism of bacterial killing derived from examination of polymorphisms in the efagin operon. The genes for efagin are clustered in the chromosome of E. faecalis and are highly conserved, indicating that it is under selection and is important to the biology of that species. However, the 3’ end of one of the genes associated with efagin expression is unusually polymorphic. This polymorphism was observed to co-vary with polymorphisms in another region of the E. faecalis chromosome that encodes the E. faecalis homolog of wall teichoic acid, termed “enterococcal polysaccharide antigen” or EPA. Experimentally, we found that the efagin encoded by one strain of E. faecalis is not effective against the producing strain, but is effective against other strains of E. faecalis with different EPA content. Structural modeling showed that the polymorphic efagin gene within was related to that encoding a tail fiber of a Listeria phage that bound the listeria cell wall polysaccharide. Moreover, the polymorphisms occurring within the efagin could be mapped to polysaccharide binding sites within the listeria phage tail fiber. From comparing genome sequences of thousands of E. faecalis strains, we believe that there are 6-8 natural variations in efagins with alterations in the cell wall carbohydrates they target. That efagins target non-homologous strains indicates they play a natural role in defense. That they target the cell wall polysaccharide of other E. faecalis strains suggests that the defense is against ecological displacement by the other E. faecalis lineages. That the activity extends to some other enterococcal species indicates that this defense may be somewhat broader than only against other E. faecalis strains. By TEM, efagins appear to be phage tail-like particles of ~ 160 nm in contour length. Other evolved phage-derived antibacterial factors have been described from a few other types of bacteria, but none are closely related to efagins, and so far, no other bacterial species has been identified where these elements are part of the core chromosome of that species.

Poster Session

Alhanouf Aljahdali

Poster presentation: Fusidic acid derivatization to enhance gram-negative potency

Alhanouf Aljahdali, Lucius Chiaraviglio, Ramchandra Donthiri, Yoon-Suk Kang, Simone Silva, Yuhan Wang, George O’Doherty* and James Kirby*

Natural products have been a rich resource for development of medical treatments. The high yield of natural products with medicinal bioactive properties is likely related to evolutionary pressures that have already optimized structures for potent physiological effects. Fusidic acid, originally isolated from Fusidium coccineum in 1960, exhibits potent activity bacteriostatic antimicrobial activity against gram-positive bacteria such as Staphylococcus aureus. It functions by targeting elongation factor G (EF-G) from the bacterial 70S ribosome and thereby inhibiting ribosomal translocation and associated protein synthesis . The objective of our study was to explore structure-activity relationships within the fusidic acid natural product scaffold to identify fusidic acid structural analogs with an enhanced activity against gram-negative ESKAPE pathogens. Native lack of gram-negative activity appears to be largely related to RND efflux pump exclusion presumptively based on hydrophic interactions. Our design principle was to first probe the fusidic acid structure for sites that are amenable for substitution in the A, C and D rings. Based on our initial findings, we then explored the effects of installation of a positive charge on the A-ring (e.g., protonated amines) on antibacterial activity. Thus far we identified a large number of modifications of the A-ring which preserve gram-positive antibacterial activity. However, significant gains in gram-negative activity have not yet been obtained.

-

Presenting author: Alhanouf Aljahdali, PhD

Post doctoral associate, Pathology, BIDMC

she/her/hers

Kathleen P. Davis

Poster presentation: Identifying underutilized antibiotic combinations for MRSA bacteremia

Kathleen Davis, Laura McDermott, Bree Aldridge, David Snydman

Methicillin-resistant Staphylococcus aureus (MRSA) bacteremia remains a serious clinical challenge. Combination therapy is mainly used against MRSA bacteremia in cases of initial treatment failure or for complicated, high-risk infections. However, frequent treatment failures and rising resistance rates will likely increase the need for combination therapy, and the nuances of combination selection are not well understood. Using a large panel of MRSA bacteremia isolates with a variety of resistance profiles, we tested eleven pairwise combinations: daptomycin and vancomycin each in combination with gentamicin, rifampicin, cefazolin, and oxacillin, as well as ceftaroline in combination with daptomycin, gentamicin, and rifampicin. We measured combination interaction, as well as the fold-change in daptomycin, vancomycin, or ceftaroline required to reach 95% growth inhibition (IC95) when a second antibiotic was added. Daptomycin + ceftaroline and ceftaroline + rifampicin demonstrated the best and most frequent occurrences of synergy. Addition of ceftaroline or cefazolin always lowered the amount of daptomycin required to reach IC95, and addition of daptomycin or rifampicin always lowered the amount of ceftaroline required to reach IC95. Our results suggest these three combinations in particular need further in vivo evaluation, and that different beta-lactams perform differently in combination.

-

Presenting author: Kathleen P. Davis, PhD

Postdoctoral Scholar, Tufts University Department of Molecular Biology and Microbiology

she/her/hers

Matthew M. Hernandez

Poster presentation: Characterization of a KPC-producing Aeromonas hydrophila from multiple anatomic compartments

Matthew M. Hernandez, Matthew Gwiazdon, Howard S. Gold, Barbra M. Blair, Patrick S. Gordon, Dana E. Pepe, Jamie M. McGloin, Thea Brennan-Krohn, Ramy Arnaout, Stefan Riedel, James E. Kirby

Background: The Aeromonas hydrophila group of bacteria are infectious threats to fish and aquaculture systems. However, Aeromonas species can also cause both intestinal and extra-intestinal infections in patients. Here, we characterize a multidrug resistant Aeromonas isolate recovered from the respiratory tract and intraabdominal space of a patient with prior, prolonged carbapenem exposure for complications after liver transplant.

Methods: Bacteria were cultured from bronchoalveolar lavage (BAL) and peritoneal abscess specimens and identified by MALDI-TOF (Bruker). Antimicrobial susceptibility testing (AST) was performed by microbroth dilution or disk diffusion methods in accordance with CLSI guidelines. Testing for carbapenamase resistance genes in isolates and rectal surveillance specimen was performed by Xpert CARBA-R assay. In vitro synergy testing was performed using the Tecan D300e digital dispenser. DNA was extracted from isolates and rectal surveillance lysate for whole genome sequencing (WGS) prior to assembly, multilocus sequence typing (MLST), and mapping of resistance genes.

Results: BAL and peritoneal abscess specimens collected one week apart grew Gram negative rods which identified as Aeromonas hydrophila group. AST revealed both isolates were resistant to all beta-lactams and beta-lactam/lactamase-inhibitor combinations. Both isolates and rectal surveillance specimen harbored KPC resistance by molecular detection. While minimum inhibitory concentration for aztreonam was >64μg/mL for both isolates, this reduced to 0.5μg/mL in combination with 4μg/mL avibactam. Sequencing of isolates generated >4Mb genomes that identified as Aeromonas hydrophila by MLST and revealed multiple beta-lactamases including KPC-2, OXA-726, and cphA2. Reads recovered from sequencing of rectal surveillance lysate also aligned with Aeromonas species.

Conclusions: We present a multidrug resistant Aeromonas hydrophila isolated from a patient with complicated respiratory and intraabdominal infections. WGS demonstrates the presence of diverse beta-lactamases including KPC-2 which has become increasingly reported among Aeromonas isolates. We also show evidence of enteric colonization by the isolate as well as presence of detectable KPC resistance. Together, these findings highlight the pathogenicity of Aeromonas and its potential to serve as a diverse reservoir for antimicrobial resistance.

---

Presenting author: Matthew M. Hernandez, MD, PhD

Medical Microbiology Fellow, Department of Pathology, Beth Israel Deaconess Medical Center

he/him/his

Yanqin Huang

Poster presentation: Efficacy of Aminoglycosides Apramycin and Plazomycin Against Mycobacterium abscessus

Yanqin Huang, PhD, Katherine Truelson, PhD candicate, Isabella A. Stewart1, PhD candicate, James E. Kirby, MD

Background: Apramycin, an aminoglycoside used in veterinary medicine, does not appear to cause ototoxicity, and both apramycin and plazomycin may offer reduced renal toxicity. Treatment of Mycobacterium abscessus infections, a key cause of chronic pulmonary disease, often makes use of amikacin in combination with other antimicrobials. Ototoxicity and renal toxicity from prolonged use of amikacin is common. Replacing amikacin with alternative aminoglycosides in these combinations may offer more acceptable alternative. Therefore, we examined the activity spectrum of apramycin and plazomycin against M. abscessus clinical isolates.

Methods:MICs for apramycin, amikacin, tobramycin, plazomicin were assessed in 40 M. abscessus isolates. Using a highly accurate and precise inkjet printing, checkboard synergy test, we tested the activity of apramycin and amikacin alone and in combination with linezolid, clofazimine, tigecycline against three isolates. Time-kill assays gauged apramycin/amikacin at 1xMIC, 2xMIC, 4xMIC with clofazimine at 2xMIC for one isolate.

Results:Susceptibility rates: apramycin (65%), amikacin (27.5%), tobramycin (2.5%), plazomicin (0%) (Figure 1). Checkboard assays: apramycin combos had FICs 1.5-3 (mode 1.5), amikacin combos ranged 1.5-5.5 (mode 3) (Figure2). Time-kill: clofazimine (2xMIC) showed minimal effect. However, apramycin (1xMIC, 2xMIC, 4xMIC) yielded 3.31, 3.67, 4.80 log10CFU reductions; amikacin (1xMIC, 2xMIC, 4xMIC) produced 0.42, 0.89, 2.15 log10CFU reductions vs. clofazimine 2xMIC monotherapy (Figure 3).

Conclusions:Apramycin displayed higher potency against M. abscessus than amikacin, while offering similar in vivo exposure based on pharmacokinetics, while plazomycin was inactive. Apramycin may offer a safer, more effective M. abscessus treatment option, and therefore merits further exploration.

-

Presenting author: Yanqin Huang, PhD

Postdoc, BIDMC, Harvard Medical School

she/her/hers

David Jimenez

Poster presentation: Novel High-Throughput Automated Antimicrobial Susceptibility Testing of Cefiderocol On The Selux Next-Generation Phenotyping System

D. Jimenez 1, K. Baker 1, B. Spears 1, K. Robets 1, K. Flentie 1, G. Michaels 1, E. Stern 1

1 Selux Dx , Charlestown, MA

Background: Adaptable, efficient, and comprehensive antimicrobial susceptibility testing (AST) systems are a fundamental component of effective antibiotic treatment. Multi-drug resistant (MDR) bacteria continue to emerge at alarming rates, requiring continuous innovation and development of new antibiotics. Widespread adoption of new antibiotics is often hindered by the absence of these drugs on automated AST systems, requiring labs to use slow and laborious manual methods. Cefiderocol (FDC), the only FDA-approved siderophore cephalosporin critical for treating carbapenem-resistant gram-negative infections, has been particularly difficult to implement in part due to its requirement for iron-depleted media. Selux’s next-generation phenotyping (NGP) system has a unique 384-well AST panel, enabling broad antibiotic menus and high adaptability to incorporate new life-saving antibiotics. Here we assess the accuracy of Cefiderocol AST using our automated system and in media containing varied iron content.

Methods: For FDC susceptibility, AST was performed using the Selux AST system and compared to CLSI broth microdilution (BMD) reference method in either cation-adjusted Muller Hinton broth (CAMHB) or its Iron depleted counterpart (ID-CAMHB). Cefiderocol was tested in ID-CAMHB across 42 total CDC AR-Bank.
Reference MICs were determined according to CLSI guidelines and Selux MICs were determined as the highest dilution producing signal <30% relative to the untreated control from the Selux system. Essential agreement (EA) and categorical agreement (CA) relative to ID-CAMHB were determined according the FDA breakpoints. Comparison of media type within each method was accessed by EA and CA.

Results: The Selux NGP system produced MICs to Cefiderocol with an average accuracy of 100% EA and 98% CA in ID-CAMHB. The use of CAMHB on the Selux NGP system showed an average EA and CA accuracy of 93% and 95% respectively when compared to BMD with ID-CAMHB reference method results.

Conclusion: The Selux NGP system’s ability to incorporate newly approved antibiotics, such as Cefiderocol, will increase the availability of essential novel agents to frontline antibiotic stewards. Selux’s flexibility combined with broad antibiotic menu and rapid AST , constitutes a valuable tool to deliver effective antibiotic therapies.

-

Presenting author: David Jimenez B.S.

Selux Diagnostics; Senior Research Associate- Microbiology

he/him/his

Greg Medlock

Poster presentation: VE707, a Defined Live Biotherapeutic Product for Prevention of Infection by Multidrug-Resistant Gram-Negative Bacteria

Gregory Medlock, Cintia Felix, Wajd Alsharif, Louis Cornacchione, Matthew Schinn, Sue Bedard-Shurtleff, Jason Norman, Jeremiah Faith, Ed Kuijper, Bernat Olle, and Silvia Caballero

Background: Infections with multidrug-resistant organisms (MDRO) are increasing at an alarming rate in hospitals worldwide. MDRO infections are often preceded by asymptomatic intestinal colonization and proliferation by the MDRO. Treatments such as non-absorbable antibiotics and fecal microbiota transplantation (FMT), which decrease the abundance of MDRO in the intestines, have shown efficacy at preventing MDRO infection. Despite the success of FMT at reducing intestinal MDRO abundance without leading to resistance, FMT composition and efficacy is variable and its safety profile questionable. This highlights the need for a defined microbiome-based product with robust efficacy and standardized manufacturing.

Methods: VE707 is a defined live biotherapeutic product (LBP) consisting of a consortium of bacterial strains that reduce intestinal carriage of carbapenem-resistant and extended-spectrum beta-lactamase-producing Klebsiella pneumoniae (Kpn) and Escherichia coli (Eco). Using a top-down approach, we characterized fecal material from healthy individuals for their ability to suppress Kpn and Eco ex vivo and in vivo, then identified donor material enriched for activity against both pathogens. Next, we used in vitro, in vivo, and bioinformatic approaches to design defined LBPs using the bacterial strains from this donor based on their anti-MDRO activity. We evaluated the ability of these LBPs to reduce Kpn and Eco abundance in a mouse co-colonization model.

Results: Of 94 LBPs designed and evaluated which each consisted of between 7 and 50 strains, VE707 showed the greatest activity, as demonstrated by a > 3-log10 reduction in Kpn and Eco in stool (Figure 1; p = 0.0022). Furthermore, VE707 was active in vitro against a panel of 40 MDR Kpn and Eco clinical isolates (> 2-log10 reduction in Kpn and Eco growth, p < 0.05). We have developed a co-culture process to enable commercially feasible manufacturing of VE707 and found that VE707 produced via co-culture has in vivo anti-MDRO activity equivalent to VE707 produced via monoculture (Figure 1; > 3-log10 reduction in Kpn and Eco in stool, p = 0.0022).

Conclusions: Our results show that VE707, a defined bacterial consortium with broad anti-MDRO activity, is successful at decolonizing Kpn and Eco and can be manufactured efficiently with a co-culture process.

-

Presenting author: Greg Medlock

Director, Vedanta Biosciences

he/him/his

Michelle Naegeli

Poster presentation: Activity of novel beta-lactam/beta-lactamase inhibitor combinations against carbapenemase-producing Enterobacterales

Michelle Naegeli, Shade Rodriguez, Thea Brennan-Krohn

Background: Combinations of beta-lactam (BL) antibiotics and beta-lactamase inhibitors (BLIs) are used to treat gram-negative bacterial infections. New diazabicyclooctane (DBO) BLIs, in addition to inhibiting beta-lactamases (including serine carbapenemases), also have direct antimicrobial activity resulting from penicillin-binding protein 2 (PBP2) inhibition. However, resistance to PBP2 inhibitors develops rapidly during drug exposure, potentially limiting clinical utility. We plan to test baseline and treatment-emergent resistance to novel combinations of BLs and DBO BLIs in order to identify combinations that are least susceptible to the emergence of resistance. In the first phase of this work, we evaluated avibactam, a DBO used clinically in the combination ceftazidime-avibactam, which has modest direct activity against Enterobacterales species.

Methods: Minimal inhibitory concentrations (MICs) of avibactam and 5 BLs with different PBP targets (aztreonam, mecillinam, meropenem, ceftazidime and cefepime) were assessed for 46 carbapenemase-producing Enterobacterales (CRE) strains. Checkerboard array synergy testing was performed using a high-throughput inkjet printer-assisted method to evaluate the effect of avibactam in combination with each BL.

Results: With the addition of avibactam (AVI) at 4 μg/mL, the MIC50 of ceftazidime fell from >16 to 2 ug/mL, mecillinam from >32 to 1 ug/mL, aztreonam from >16 to 1 ug/mL, cefepime from >16 to ≤0.25 ug/mL, and meropenem from 4 to ≤0.06 ug/mL. Among metallo-beta-lactamase (MBL)-producing strains (n = 7), synergy with AVI was only observed with aztreonam (6/7 strains; expected as aztreonam is not hydrolyzed by MBLs) and mecillinam (3/7 strains). The MIC50 of AVI alone for all strains was 32 ug/mL.

Conclusion: The addition of AVI significantly reduced MICs of 5 different BLs against serine carbapenemase-producing CRE isolates, with the most significant drops in MIC seen with meropenem, cefepime, and mecillinam. Reduction in MIC due to inhibition of serine carbapenemases is expected, but the reason for the differential synergistic effect is not known and may result from dual inhibition of PBP2 by AVI and by BLs that bind PBP2 (mecillinam and, to a lesser extent, meropenem and cefepime). Synergy between mecillinam and avibactam in MBL-producing strains may also involve direct AVI activity. In future work we will test newer, more potent DBOs (zidebactam, durlobactam), and will evaluate rates of treatment-emergent resistance to the combinations.
 

-

Presenting author: Michelle Naegeli, MD

Postdoctoral Research Fellow at the Department of Pathology, Beth Israel Deaconess Medical Center & Harvard Medical School

she/her/hers

Danielle Peterson

Poster presentation: Keynome ID: an alignment-free algorithm for rapid species identification on whole blood

Danielle Peterson¹, Defne Surujon¹, Connor Parish¹, Mohamad Sater¹, Miriam Huntley¹

¹Day Zero Diagnostics- Boston, Massachusetts

The current gold standard for bloodstream infection (BSI) diagnostics requires culturing for pathogen identification and antibiotic susceptibility testing (AST). A major limitation for culture-based approaches is that it takes far too long to get to an actionable result. Since the mortality rate for BSI patients is alarmingly high (up to 82% for septic shock patients within the first 12 hours), initial intervention is often done with broad-spectrum antibiotics, and the culture results are used for subsequent course-correction when they become available. A rapid diagnostic can improve antibiotic stewardship by reducing the overuse of broad-spectrum antibiotics or by shifting to more effective targeted antibiotics when unusual resistance is present. Here, we present Keynome ID, an algorithm that identifies the species present in whole blood samples processed with Blood2Bac, a pathogen ultra-high enrichment technology, and sequenced on the Oxford Nanopore Technologies (ONT) platform. Briefly, the sequencing data is filtered for quality, and human-derived reads are removed. The remaining reads are assembled. The assemblies are then taxonomically scored and binned using our curated and quality controlled database, MicroRefDB. For each species detected in a sample, Keynome ID outputs a score that estimates the breadth of genomic coverage, which is then converted to a presence/absence call based on pre-set cutoff values. Keynome ID is an alignment-free method, which is an improvement on three main disadvantages of alignment-based methods: 1. without a priori knowledge of what species are present in the sample, alignment-based methods can require a lot of compute resources 2. alignment-based methods are prone to noise and false positive calls due to cross-mapping between closely related species that share genomic regions, and 3. Finding an appropriate reference genome for accurate assessment of coverage can be challenging, especially when there is a lot of within-species variability or in the case of less common species. Keynome ID is both species-agnostic, and has high specificity, while having a rapid turn-around-time of <30 minutes. Moreover, Keynome ID is able to output a whole genome assembly, which is critical for comprehensive AST predictions. We demonstrate the performance of Keynome ID on a panel of 30 pathogens, including bacterial and fungal species, spiked into whole blood. When paired with Blood2Bac, Keynome ID enables rapid pathogen identification, and has the potential to improve antibiotic stewardship practices in a clinical setting.

-

Presenting author: Danielle Peterson, BS

Bioinformatician, Day Zero Diagnostics

she/her/hers

Anusha Shukla

Poster presentation: Synergistic Activity of Tobramycin Combined with Histone Demethylase Inhibitor GSK-J4 against Burkholderia cepacia Complex Pathogens

Anusha Shukla, Thea Brennan-Krohn

Background: Burkholderia cepacia complex (Bcc) is a significant cause of morbidity and mortality in people with cystic fibrosis (CF). Bcc’s intrinsic resistance to many antibiotics and its ability to rapidly acquire additional resistance mechanisms limit treatment options. Identification of existing known bioactive compounds that have activity against Bcc either alone or as antibiotic potentiators could dramatically shorten time to clinical availability, particularly for drugs that are already FDA-approved. A high-throughput screening identified GSK-J4, a histone demethylase inhibitor targeting Jumanji-C domain-containing proteins, as a synergistic hit with Tobramycin and follow-up experiments confirmed the efficacy of this combination.

Methods: A high throughput screening was performed in duplicate with libraries of known bioactive compounds at ICCB-Longwood Screening Facility (Harvard Medical School, Boston, MA). The HP D300 digital dispenser was used to perform an inkjet printer-based checkboard synergy assay to test the GSK-J4 and tobramycin combination at varying concentrations under standard antimicrobial susceptibility testing conditions. A time-kill assay was also performed to evaluate the time course of bacterial growth and bactericidal activity of the combination.

Results: The combination of GSK-J4 and Tobramycin (TOB) showed potentially clinically relevant synergy when tested against Bcc strains Burkholderia cenocepacia K562 and Burkholderia multivorans CGD1. Similar synergistic activity was also observed for the combination of GSK-J4 with one of amikacin, apramycin, gentamycin, streptomycin and nourseothricin, suggesting a class effect. Other histone demethylase inhibitors were also tested in combination with TOB and one other hit, PBIT, was discovered. GSK-J4 had no activity against strains of Acinetobacter baumannii, Klebsiella pneumonia, Pseudomonas aeruginosa and Escherichia coli by itself or combined with tobramycin, but was effective on its own against an Escherichia coli tolC mutant which lacks its main efflux pump.

Conclusions: The combination of GSK-J4 and TOB has specific antimicrobial activity against Bcc strains and could be a potential therapy against Burkholderia infections in Cystic Fibrosis patients. Furthermore, evaluating the active sites of targets of GSK-J4 and PBIT in humans and cross-referencing with Burkholderia genomes could provide insight into a potential target for future drug development.

Conflict of Interest Statement: The HP D300 digital dispenser and associated consumables were provided by Tecan. Tecan had no role in study design, data collection/interpretation, or poster preparation.

Funding Statement: TBK was supported by a NIAID career development award (1K08AI132716) and by NIAID award R21 AI156039.

-

Presenting author: Anusha Shukla

Research Assistant, Beth Israel Deaconess Medical Center Department of Pathology

she/her/hers

Matthew E. Turner

Poster presentation: Identification and Antibiotic Resistance Profiling of Bacterial Pathogens Directly from Whole Human Blood Utilizing Blood2Bac

Matthew E. Turner¹, Paul Knysh¹, Jason Wittenbach¹, Defne Surujon¹, Ian Herriott¹, Emily MacLeod¹, Cabell Maddux¹, Cristina Rushton¹, Danielle Peterson1, Mohamad Sater¹, Linnea Sahlberg¹, Kaitlin Woodruff¹, Claire Zimmerman¹, Jenna McFaul1, Anna Early1, Nicole Billings1, Miriam H. Huntley¹, Manoj Nair¹, Douglas S. Kwon¹,²

¹Day Zero Diagnostics- Boston, Massachusetts, ²Ragon Institute of MGH, MIT, and Harvard- Cambridge, Massachusetts

Bloodstream infections (BSI) are a leading cause of patient mortality with early diagnosis and pathogen identification key to improving clinical outcomes. However, longer turn-around times associated with culture-based diagnostics limit the ability of clinical labs to provide timely information to clinicians. Moreover, delayed treatment for patients significantly impacts clinical outcomes with septic shock mortality rates increasing by 8% per hour without appropriate antibiotics. Day Zero Diagnostics has developed Blood2Bac™, a culture-free, pathogen-agnostic technology which enriches bacterial pathogens directly from whole human blood and utilizes bacterial whole genome sequencing and proprietary algorithms to provide sensitive identification of pathogens and antimicrobial resistance (AMR) information at single digit CFU/mL levels. Performance assessment of the Blood2Bac™ process made across 26 priority BSI species demonstrated analytical sensitivity at or below 5 CFU/mL concentration with average genomic coverage of 99.14% for 23 out of 26 of the tested strains. The ability to discriminate between individual species was demonstrated within 5 sets of polymicrobial samples, each containing two different priority BSI species. Additionally, assessment of Blood2Bac™ and the Keynome® ID algorithm for species identification was made on a blinded panel of 40 distinct strains provided through the IHMA biobank with 97.5% accuracy (positive ID for 39/40 blinded organisms). To test the ability of the Keynome® gAST algorithm to determine antibiotic susceptibility profiles of pathogens directly from blood, we tested 50 bacterial strains spiked into whole blood at 5.5-21.5 CFU/mL levels and demonstrated Categorical Agreement of 91.3% with phenotypic AST. As a whole, these results demonstrate the sensitivity and accuracy of Blood2Bac™ for the identification and AMR profiling of BSI pathogens in the single digit bacterial CFU/mL range direct from whole human blood.

-

Presenting author: Matthew E. Turner, PhD

Research Scientist II, Day Zero Diagnostics

he/him/his

Ziming Wei

Poster presentation: Deep learning for antibiotic discovery in Mycobacterium Tuberculosis

Ziming Wei, Yasha Ektefaie, Dereje Negatu, Bree Aldridge, Thomas Dick, Inna Krieger, James Sacchettini, Marinka Zitnik, Maha Reda Farhat

M. tuberculosis (Mtb) is the leading cause of death by infectious disease worldwide. Increasing rates of antibiotic resistance in Mtb threaten to undo decades of progress in fighting the disease. Though there is a need for new Mtb antibiotics, traditional methods of antibiotic discovery are slow and costly. Machine learning has been shown to accurately predict antibiotic effects of compounds in-sillico and promises to accelerate antibiotic discovery. However, the potential of ML for Mtb antibiotic discovery is unknown. Here we train graph neural networks and transformers on the dataset with 61,896 Mtb compounds and their inhibitory effect on Mtb growth, as determined by luminescence and resazurin readouts. We found these models achieve high sensitivity and specificity (> 0.75) on Mtb compound inhibitory effect prediction. We demonstrate these models can generalize to unseen classes of compounds and can accurately identify existing Mtb antibiotics from a library of compounds. We deploy these models and identify a list of compounds with predicted high Mtb inhibitory effect. Our results suggest that ML can accelerate Mtb antibiotic discovery by more specifically identifying compounds of interest for downstream experimental validation.

-

Presenting author: Ziming Wei

Graduate Research Assistant, Department of Biomedical Informatics, Harvard Medical School

he/him/his

Kelsey Wheeler

Poster presentation: Mucin Glycans as Inhibitors of Enteric Pathogen Virulence

Kelsey M. Wheeler, Michaela A. Gold, Corey Stevens, Agnes A. Walsh, Karsten Tedin, Gerardo Cárcamo-Oyarce, Jeongming Song, Marcus Fulde, Rachel Hevey, Katharina Ribbeck

Salmonella enterica serovar Typhimurium (S. Typhimurium) is a dangerous, food-borne bacterium that can invade the cells of the intestinal tract and cause severe gastroenteritis. Here, we demonstrate that exposure of S. Typhimurium to the glycoprotein mucin, the main component of the natural mucus barrier, reduces infection of cultured intestinal epithelial cells. We identify complex mucin sugars (glycans) as a biochemical signal that downregulates multiple virulence genes via a central regulatory cascade. This work highlights the role of the healthy mucosal environment as a natural anti-infective signal for S. Typhimurium and will motivate the development of mucin-inspired therapies to treat enteric pathogens.

-

Presenting author: Kelsey M Wheeler, PhD

Research Scientist, MIT Department of Biological Engineering

she/her/hers

 

Liubov Yakovlieva

Poster presentation: Mucin-inspired strategies to attenuate virulence of pathogen Gardnerella vaginalis.

Liubov Yakovlieva, C. Werlang, S. Brucks, J. Sahoo, A. Bergerat-Thompson, C. Mitchell, D. Kaplan, L. Kiessling, K. Ribbeck.

Bacterial vaginosis (BV) is a common dysbiotic vaginal condition in women of reproductive age. Besides significant discomfort, it can lead to severe gynecologic and obstetric sequelae, including miscarriage, preterm birth, and enhanced acquisition of HPV and HIV. BV is associated with high relapse rates following antibiotic treatment, with ~80% of people suffering a recurrence within a year.

Microbiologically, BV is characterized by a decline in protective Lactobacillus and rapid outgrowth of a diverse anaerobic community. One key pathogen associated with 90% of BV cases is Gardnerella vaginalis. This bacterium employs a wide range of virulence factors that exacerbate the BV condition and drive inflammation and epithelial tissue damage. These pathogenic traits include 1) secretion of mucus degradative enzymes, 2) production of cytotoxin vaginolysin to extract nutrients from the host cells, and 3) propensity to form multispecies biofilms that resist antibiotic treatment.

In the Ribbeck lab, we leverage the ability of native mucus barriers to attenuate the virulence of pathogens and utilize mucus-derived factors as potential therapeutic solutions to mitigate infections. In this poster, I will share our efforts in identifying and applying bioactive mucin-derived glycans to specifically target the virulence of Gardnerella vaginalis. We aim to narrow down glycans that can prevent and disrupt G. vaginalis biofilms, downregulate toxin production, and rescue epithelial cells from killing. We prioritize host- and Lactobacillus-safe mucin glycans that can serve as a stand-alone intervention or in combination with antibiotic or probiotic treatments. With this, we envision the development of much-needed alternative therapeutic options to prevent and combat bacterial vaginosis effectively.

-

Presenting author: Liubov Yakovlieva, Ph.D.

Postdoctoral Associate, Ribbeck Lab, MIT

she/her/hers